Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
RSC Med Chem ; 14(12): 2509-2534, 2023 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-38107174

RESUMEN

Isoquinoline-enrooted organic small-molecules represent a challenging molecular target in the organic synthesis arsenal attributed to their structural diversity and therapeutic importance. Into the bargain, isoquinolines are significant structural frameworks in modern medicinal chemistry and drug development. Consequently, synthetic organic and medicinal chemists have been intensely interested in efficient synthetic tactics for the sustainable construction of isoquinoline frameworks and their derivatives in enantiopure or racemic forms. This review accentuates an overview of the literature on the modern synthetic approaches exploited in synthesising isoquinolines and their core embedded heterocyclic skeletons from 2021 to 2022. In detail, the methodologies and inspected pharmacological studies for the array of diversely functionalized isoquinolines or their core-embedded heterocyclic/carbocyclic structures involving the introduction of substituents at C-1, C-3, and C-4 carbon and N-2 atom, bond constructions at the C1-N2 atom and C3-N2 atom, and structural scaffolding within isoquinoline compounds have been reviewed. This intensive study highlights the need for and relevance of relatively unexplored bioisosterism employing isoquinoline-based small-molecules in drug design.

2.
PLoS One ; 17(8): e0270273, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35925919

RESUMEN

Chronic hepatitis B virus (HBV) infection is characterized by the presence of high circulating levels of non-infectious lipoprotein-like HBV surface antigen (HBsAg) particles thought to contribute to chronic immune dysfunction in patients. Lipid and metabolomic analysis of humanized livers from immunodeficient chimeric mice (uPA/SCID) revealed that HBV infection dysregulates several lipid metabolic pathways. Small molecule inhibitors of lipid biosynthetic pathway enzymes acetyl-CoA carboxylase (ACC), fatty acid synthase, and subtilisin kexin isozyme-1/site-1 protease in HBV-infected HepG2-NTCP cells demonstrated potent and selective reduction of extracellular HBsAg. However, a liver-targeted ACC inhibitor did not show antiviral activity in HBV-infected liver chimeric mice, despite evidence of on-target engagement. Our study suggests that while HBsAg production may be dependent on hepatic de novo lipogenesis in vitro, this may be overcome by extrahepatic sources (such as lipolysis or diet) in vivo. Thus, a combination of agents targeting more than one lipid metabolic pathway may be necessary to reduce HBsAg levels in patients with chronic HBV infection.


Asunto(s)
Hepatitis B Crónica , Hepatitis B , Animales , Antivirales/metabolismo , Antivirales/farmacología , Antivirales/uso terapéutico , ADN Viral/metabolismo , Antígenos de Superficie de la Hepatitis B/metabolismo , Virus de la Hepatitis B/genética , Hepatitis B Crónica/tratamiento farmacológico , Lípidos/uso terapéutico , Ratones , Ratones SCID
3.
Hepatol Commun ; 6(9): 2298-2309, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35735253

RESUMEN

Dysregulated hepatocyte lipid metabolism is a hallmark of hepatic lipotoxicity and contributes to the pathogenesis of nonalcoholic steatohepatitis (NASH). Acetyl CoA carboxylase (ACC) inhibitors decrease hepatocyte lipotoxicity by inhibiting de novo lipogenesis and concomitantly increasing fatty acid oxidation (FAO), and firsocostat, a liver-targeted inhibitor of ACC1/2, is under evaluation clinically in patients with NASH. ACC inhibition is associated with improvements in indices of NASH and reduced liver triglyceride (TG) content, but also increased circulating TG in subjects with NASH and preclinical rodent models. Here we evaluated whether enhancing hepatocyte FAO by combining ACC inhibitors with peroxisomal proliferator-activated receptor (PPAR) or thyroid hormone receptor beta (THRß) agonists could drive greater liver TG reduction and NASH/antifibrotic efficacy, while ameliorating ACC inhibitor-induced hypertriglyceridemia. In high-fat diet-fed dyslipidemic rats, the addition of PPAR agonists fenofibrate (Feno), elafibranor (Ela), lanifibranor (Lani), seladelpar (Sela) or saroglitazar (Saro), or the THRb agonist resmetirom (Res), to an analogue of firsocostat (ACCi) prevented ACCi-induced hypertriglyceridemia. However, only PPARα agonists (Feno and Ela) and Res provided additional liver TG reduction. In the choline-deficient high-fat diet rat model of advanced liver fibrosis, neither PPARα (Feno) nor THRß (Res) agonism augmented the antifibrotic efficacy of ACCi. Conclusion: These data suggest that combination therapies targeting hepatocyte lipid metabolism may have beneficial effects on liver TG reduction; however, they may not be sufficient to drive fibrosis regression.


Asunto(s)
Fenofibrato , Hipertrigliceridemia , Enfermedad del Hígado Graso no Alcohólico , Acetatos , Acetil-CoA Carboxilasa , Animales , Fenofibrato/farmacología , Humanos , Cirrosis Hepática/inducido químicamente , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , PPAR alfa/uso terapéutico , Ratas , Triglicéridos/uso terapéutico
4.
Endocrinology ; 162(7)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33765118

RESUMEN

Circulating branched chain amino acid (BCAA) levels are elevated in obese humans and genetically obese rodents. However, the relationship of BCAAs to insulin resistance in diet-induced obese mice, a commonly used model to study glucose homeostasis, is still ill-defined. Here we examined how high-fat high-sucrose (HFHS) or high-fat diet (HFD) feeding, with or without BCAA supplementation in water, alters the metabolome in serum/plasma and tissues in mice and whether raising circulating BCAA levels worsens insulin resistance and glucose intolerance. Neither HFHS nor HFD feeding raised circulating BCAA levels in insulin-resistant diet-induced obese mice. BCAA supplementation raised circulating BCAA and branched-chain α-keto acid levels and C5-OH/C3-DC acylcarnitines (AC) in muscle from mice fed an HFHS diet or HFD, but did not worsen insulin resistance. A set of short- and long-chain acyl CoAs were elevated by diet alone in muscle, liver, and white adipose tissue (WAT), but not increased further by BCAA supplementation. HFD feeding reduced valine and leucine oxidation in WAT but not in muscle. BCAA supplementation markedly increased valine oxidation in muscle from HFD-fed mice, while leucine oxidation was unaffected by diet or BCAA treatment. Here we establish an extensive metabolome database showing tissue-specific changes in mice on 2 different HFDs, with or without BCAA supplementation. We conclude that mildly elevating circulating BCAAs and a subset of ACs by BCAA supplementation does not worsen insulin resistance or glucose tolerance in mice. This work highlights major differences in the effects of BCAAs on glucose homeostasis in diet-induced obese mice versus data reported in obese rats and in humans.


Asunto(s)
Aminoácidos de Cadena Ramificada/administración & dosificación , Glucemia/metabolismo , Dieta/efectos adversos , Resistencia a la Insulina/fisiología , Metabolómica , Obesidad/metabolismo , Aminoácidos de Cadena Ramificada/sangre , Aminoácidos de Cadena Ramificada/metabolismo , Animales , Dieta Alta en Grasa , Sacarosa en la Dieta/administración & dosificación , Suplementos Dietéticos , Femenino , Intolerancia a la Glucosa/sangre , Homeostasis/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Obesidad/etiología , Oxidación-Reducción
5.
J Hepatol ; 73(4): 896-905, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32376414

RESUMEN

BACKGROUND & AIMS: Non-alcoholic steatohepatitis (NASH) is a chronic liver disease characterized by hepatic lipid accumulation, inflammation, and progressive fibrosis. Acetyl-CoA carboxylase (ACC) catalyzes the rate-limiting step of de novo lipogenesis and regulates fatty acid ß-oxidation in hepatocytes. ACC inhibition reduces hepatic fat content and markers of liver injury in patients with NASH; however, the effect of ACC inhibition on liver fibrosis has not been reported. METHODS: A direct role for ACC in fibrosis was evaluated by measuring de novo lipogenesis, procollagen production, gene expression, glycolysis, and mitochondrial respiration in hepatic stellate cells (HSCs) in the absence or presence of small molecule inhibitors of ACC. ACC inhibitors were evaluated in rodent models of liver fibrosis induced by diet or the hepatotoxin, diethylnitrosamine. Fibrosis and hepatic steatosis were evaluated by histological and biochemical assessments. RESULTS: Inhibition of ACC reduced the activation of TGF-ß-stimulated HSCs, as measured by both α-SMA expression and collagen production. ACC inhibition prevented a metabolic switch necessary for induction of glycolysis and oxidative phosphorylation during HSC activation. While the molecular mechanism by which inhibition of de novo lipogenesis blocks glycolysis and oxidative phosphorylation is unknown, we definitively show that HSCs require de novo lipogenesis for activation. Consistent with this direct antifibrotic mechanism in HSCs, ACC inhibition reduced liver fibrosis in a rat choline-deficient, high-fat diet model and in response to chronic diethylnitrosamine-induced liver injury (in the absence of hepatic lipid accumulation). CONCLUSIONS: In addition to reducing lipid accumulation in hepatocytes, ACC inhibition also directly impairs the profibrogenic activity of HSCs. Thus, small molecule inhibitors of ACC may lessen fibrosis by reducing lipotoxicity in hepatocytes and by preventing HSC activation, providing a mechanistic rationale for the treatment of patients with advanced liver fibrosis due to NASH. LAY SUMMARY: Hepatic fibrosis is the most important predictor of liver-related outcomes in patients with non-alcoholic steatohepatitis (NASH). Small molecule inhibitors of acetyl-CoA carboxylase (ACC) reduce hepatic fat content and markers of liver injury in patients with NASH. Herein, we report that inhibition of ACC and de novo lipogenesis also directly suppress the activation of hepatic stellate cells - the primary cell responsible for generating fibrotic scar in the liver - and thus fibrosis. These data provide further evidence for the use of ACC inhibitors to treat patients with NASH and advanced fibrosis.


Asunto(s)
Acetil-CoA Carboxilasa/antagonistas & inhibidores , Células Estrelladas Hepáticas/metabolismo , Lipogénesis/efectos de los fármacos , Cirrosis Hepática/metabolismo , Hígado/patología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Biomarcadores/metabolismo , Línea Celular , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/patología , Humanos , Hígado/metabolismo , Cirrosis Hepática/etiología , Cirrosis Hepática/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Ratas , Ratas Wistar
6.
J Clin Invest ; 129(9): 3717-3731, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31380811

RESUMEN

Palmitic acid esters of hydroxy stearic acids (PAHSAs) are endogenous antidiabetic and antiinflammatory lipids. Here, we show that PAHSAs protect against type 1 diabetes (T1D) and promote ß cell survival and function. Daily oral PAHSA administration to nonobese diabetic (NOD) mice delayed the onset of T1D and markedly reduced the incidence of T1D, whether PAHSAs were started before or after insulitis was established. PAHSAs reduced T and B cell infiltration and CD4+ and CD8+ T cell activation, while increasing Treg activation in pancreata of NOD mice. PAHSAs promoted ß cell proliferation in both NOD mice and MIN6 cells and increased the number of ß cells in NOD mice. PAHSAs attenuated cytokine-induced apoptotic and necrotic ß cell death and increased ß cell viability. The mechanism appears to involve a reduction of ER stress and MAPK signaling, since PAHSAs lowered ER stress in NOD mice, suppressed thapsigargin-induced PARP cleavage in human islets, and attenuated ERK1/2 and JNK1/2 activation in MIN6 cells. This appeared to be mediated in part by glucagon-like peptide 1 receptor (GLP-1R) and not the G protein-coupled receptor GPR40. PAHSAs also prevented impairment of glucose-stimulated insulin secretion and improved glucose tolerance in NOD mice. Thus, PAHSAs delayed the onset of T1D and reduced its incidence by attenuating immune responses and exerting direct protective effects on ß cell survival and function.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Diabetes Mellitus Tipo 1/inmunología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Ácido Palmítico/farmacología , Ácidos Esteáricos/farmacología , Adulto , Anciano , Animales , Diabetes Mellitus Experimental/inmunología , Ésteres , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Sistema Inmunológico , Insulina/metabolismo , Islotes Pancreáticos/inmunología , Masculino , Ratones , Ratones Endogámicos NOD , Persona de Mediana Edad , Linfocitos T/inmunología
7.
Sci Rep ; 8(1): 15757, 2018 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-30361530

RESUMEN

Adipose tissue dysfunction is considered an important contributor to systemic insulin resistance and Type 2 diabetes (T2D). Recently, a novel family of endogenous lipids, palmitic acid hydroxy stearic acids (PAHSAs), was discovered. These have anti-diabetic and anti-inflammatory effects in mice and are reduced in serum and adipose tissue of insulin resistant humans. In the present study, we investigate if adipose tissue dysfunction is associated with reduced PAHSA levels in human subjects and if PAHSAs influence adipocyte differentiation. Our results show that low expression of adipocyte GLUT4 and adipocyte hypertrophy, markers of adipose tissue dysfunction, are associated with reduced expression of key enzymes for de novo lipogenesis and adipose tissue levels of PAHSAs in human subjects. We also show that GLUT4 is not only a marker of adipose tissue dysfunction, but may be causally related to the observed impairments. PAHSAs may also act locally in the adipose tissue to improve adipogenesis through a mechanism bypassing direct activation of peroxisome proliferator-activated receptor (PPARγ). The discovery of PAHSAs and our current results provide novel insights into positive effects of lipid species in adipose tissue and mechanisms by which dysfunctional adipose tissue is associated with insulin resistance and risk of developing T2D.


Asunto(s)
Ácido Palmítico/metabolismo , Ácidos Esteáricos/metabolismo , Grasa Subcutánea/fisiopatología , Células 3T3-L1 , Adipocitos/metabolismo , Adipocitos/patología , Adipogénesis , Adulto , Animales , Femenino , Silenciador del Gen , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Hipertrofia , Resistencia a la Insulina , Masculino , Ratones , PPAR gamma/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Regresión , Grasa Subcutánea/patología , Activación Transcripcional/genética
8.
Nat Rev Mol Cell Biol ; 19(10): 654-672, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30104701

RESUMEN

The cause of insulin resistance in obesity and type 2 diabetes mellitus (T2DM) is not limited to impaired insulin signalling but also involves the complex interplay of multiple metabolic pathways. The analysis of large data sets generated by metabolomics and lipidomics has shed new light on the roles of metabolites such as lipids, amino acids and bile acids in modulating insulin sensitivity. Metabolites can regulate insulin sensitivity directly by modulating components of the insulin signalling pathway, such as insulin receptor substrates (IRSs) and AKT, and indirectly by altering the flux of substrates through multiple metabolic pathways, including lipogenesis, lipid oxidation, protein synthesis and degradation and hepatic gluconeogenesis. Moreover, the post-translational modification of proteins by metabolites and lipids, including acetylation and palmitoylation, can alter protein function. Furthermore, the role of the microbiota in regulating substrate metabolism and insulin sensitivity is unfolding. In this Review, we discuss the emerging roles of metabolites in the pathogenesis of insulin resistance and T2DM. A comprehensive understanding of the metabolic adaptations involved in insulin resistance may enable the identification of novel targets for improving insulin sensitivity and preventing, and treating, T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Resistencia a la Insulina/fisiología , Animales , Trastornos del Metabolismo de la Glucosa/metabolismo , Humanos , Insulina/metabolismo , Resistencia a la Insulina/genética , Metabolismo de los Lípidos/fisiología , Hígado/metabolismo , Redes y Vías Metabólicas , Metabolómica/métodos , Obesidad , Receptor de Insulina/metabolismo , Transducción de Señal
9.
Cell Rep ; 21(4): 1021-1035, 2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-29069585

RESUMEN

Lower adipose-ChREBP and de novo lipogenesis (DNL) are associated with insulin resistance in humans. Here, we generated adipose-specific ChREBP knockout (AdChREBP KO) mice with negligible sucrose-induced DNL in adipose tissue (AT). Chow-fed AdChREBP KO mice are insulin resistant with impaired insulin action in the liver, muscle, and AT and increased AT inflammation. HFD-fed AdChREBP KO mice are also more insulin resistant than controls. Surprisingly, adipocytes lacking ChREBP display a cell-autonomous reduction in insulin-stimulated glucose transport that is mediated by impaired Glut4 translocation and exocytosis, not lower Glut4 levels. AdChREBP KO mice have lower levels of palmitic acid esters of hydroxy stearic acids (PAHSAs) in serum, and AT. 9-PAHSA supplementation completely rescues their insulin resistance and AT inflammation. 9-PAHSA also normalizes impaired glucose transport and Glut4 exocytosis in ChREBP KO adipocytes. Thus, loss of adipose-ChREBP is sufficient to cause insulin resistance, potentially by regulating AT glucose transport and flux through specific lipogenic pathways.


Asunto(s)
Adipocitos/metabolismo , Glucosa/metabolismo , Resistencia a la Insulina , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Células 3T3 , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Células Cultivadas , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Proteínas Nucleares/genética , Ácidos Palmíticos/sangre , Ácidos Esteáricos/sangre , Factores de Transcripción/genética
10.
Endocrinology ; 154(10): 3776-83, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23861377

RESUMEN

In this study, we investigated whether loss of GH receptor (GHR) signaling in postnatal skeletal muscle alters muscle mass and regenerative ability in adult mice and whether this was dependent on IGF-1 receptor (IGF-1R) signaling. To do so, we used mouse models with skeletal muscle-specific loss of GHR signaling (mGHRKO), IGF-1R and insulin receptor signaling (MKR), or both GHR and IGF-1R/insulin receptor signaling (mGHRKO/MKR). We did not find a reduction in muscle cross-sectional area, fiber type composition, or response to pathological muscle injury in male mGHRKO and mGHRKO/MKR mice when compared with control and MKR mice, respectively. This could potentially be explained by unchanged skeletal muscle Igf-1 expression in mGHRKO and mGHRKO/MKR mice relative to control and MKR mice, respectively. Furthermore, MKR and mGHRKO/MKR mice, but not mGHRKO mice, demonstrated reduced fiber fusion after cardiotoxin injection, suggesting that IGF-1, and not GH, promotes fiber fusion in adult mice. In summary, our data suggest that GHR signaling in postnatal skeletal muscle does not play a significant role in regulating muscle mass or muscle regeneration. Additionally, in our model, muscle Igf-1 expression is not dependent on GHR signaling in postnatal skeletal muscle.


Asunto(s)
Desarrollo de Músculos , Músculo Esquelético/fisiología , Receptor IGF Tipo 1/metabolismo , Receptores de Somatotropina/metabolismo , Regeneración , Transducción de Señal , Animales , Cardiotoxinas/toxicidad , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/metabolismo , Hormona del Crecimiento/metabolismo , Humanos , Insulina/metabolismo , Insulina/uso terapéutico , Resistencia a la Insulina , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Receptor IGF Tipo 1/biosíntesis , Receptor IGF Tipo 1/genética , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Receptores de Somatotropina/genética , Transducción de Señal/efectos de los fármacos
11.
PLoS One ; 7(9): e44777, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23024761

RESUMEN

BACKGROUND: Growth hormone (GH) stimulates whole-body lipid oxidation, but its regulation of muscle lipid oxidation is not clearly defined. Mice with a skeletal muscle-specific knockout of the GH receptor (mGHRKO model) are protected from high fat diet (HFD)-induced insulin resistance and display increased whole-body carbohydrate utilization. In this study we used the mGRHKO mice to investigate the role of muscle GHR signaling on lipid oxidation under regular chow (RC)- and HFD- fed conditions, and in response to fasting. METHODOLOGY/PRINCIPAL FINDINGS: Expression of lipid oxidation genes was analyzed by real-time PCR in the muscles of RC- and HFD- fed mice, and after 24 h fasting in the HFD-fed mice. Expression of lipid oxidation genes was lower in the muscles of the mGHRKO mice relative to the controls, irrespective of diet. However, in response to 24 h fasting, the HFD-fed mGHRKO mice displayed up-regulation of lipid oxidation genes similar to the fasted controls. When subjected to treadmill running challenge, the HFD-fed mGHRKO mice demonstrated increased whole-body lipid utilization. Additionally, under fasted conditions, the adipose tissue of the mGHRKO mice displayed increased lipolysis as compared to both the fed mGHRKO as well as the fasted control mice. CONCLUSIONS/SIGNIFICANCE: Our data show that muscle GHR signaling regulates basal lipid oxidation, but not the induction of lipid oxidation in response to fasting. We further demonstrate that muscle GHR signaling is involved in muscle-adipose tissue cross-talk; however the mechanisms mediating this remain to be elucidated.


Asunto(s)
Metabolismo de los Lípidos , Músculo Esquelético/metabolismo , Receptores de Somatotropina/metabolismo , Transducción de Señal , Tejido Adiposo/metabolismo , Animales , Dieta Alta en Grasa , Ayuno , Regulación de la Expresión Génica , Metabolismo de los Lípidos/genética , Lipólisis , Masculino , Ratones , Ratones Noqueados , Mitocondrias Musculares/genética , Mitocondrias Musculares/metabolismo , Oxidación-Reducción , Condicionamiento Físico Animal , Receptores de Somatotropina/genética
12.
Diabetes ; 61(1): 94-103, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22187377

RESUMEN

Growth hormone (GH) exerts diverse tissue-specific metabolic effects that are not revealed by global alteration of GH action. To study the direct metabolic effects of GH in the muscle, we specifically inactivated the growth hormone receptor (ghr) gene in postnatal mouse skeletal muscle using the Cre/loxP system (mGHRKO model). The metabolic state of the mGHRKO mice was characterized under lean and obese states. High-fat diet feeding in the mGHRKO mice was associated with reduced adiposity, improved insulin sensitivity, lower systemic inflammation, decreased muscle and hepatic triglyceride content, and greater energy expenditure compared with control mice. The obese mGHRKO mice also had an increased respiratory exchange ratio, suggesting increased carbohydrate utilization. GH-regulated suppressor of cytokine signaling-2 (socs2) expression was decreased in obese mGHRKO mice. Interestingly, muscles of both lean and obese mGHRKO mice demonstrated a higher interleukin-15 and lower myostatin expression relative to controls, indicating a possible mechanism whereby GHR signaling in muscle could affect liver and adipose tissue function. Thus, our study implicates skeletal muscle GHR signaling in mediating insulin resistance in obesity and, more importantly, reveals a novel role of muscle GHR signaling in facilitating cross-talk between muscle and other metabolic tissues.


Asunto(s)
Proteínas Portadoras/genética , Dieta Alta en Grasa/efectos adversos , Enfermedades Metabólicas/etiología , Músculo Esquelético/metabolismo , Animales , Proteínas Portadoras/metabolismo , Proteínas Portadoras/fisiología , Citoprotección/genética , Eliminación de Gen , Marcación de Gen , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Masculino , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética , Obesidad/metabolismo , Receptor Cross-Talk/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología
13.
J Clin Invest ; 121(6): 2422-6, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21555853

RESUMEN

Insulin, growth hormone (GH), and insulin-like growth factor-1 (IGF-1) play key roles in the regulation of ß cell growth and function. Although ß cells express the GH receptor, the direct effects of GH on ß cells remain largely unknown. Here we have employed a rat insulin II promoter-driven (RIP-driven) Cre recombinase to disrupt the GH receptor in ß cells (ßGHRKO). ßGHRKO mice fed a standard chow diet exhibited impaired glucose-stimulated insulin secretion but had no changes in ß cell mass. When challenged with a high-fat diet, ßGHRKO mice showed evidence of a ß cell secretory defect, with further deterioration of glucose homeostasis indicated by their altered glucose tolerance and blunted glucose-stimulated insulin secretion. Interestingly, ßGHRKO mice were impaired in ß cell hyperplasia in response to a high-fat diet, with decreased ß cell proliferation and overall reduced ß cell mass. Therefore, GH receptor plays critical roles in glucose-stimulated insulin secretion and ß cell compensation in response to a high-fat diet.


Asunto(s)
Grasas de la Dieta/farmacología , Intolerancia a la Glucosa/etiología , Glucosa/fisiología , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Obesidad/fisiopatología , Receptores de Somatotropina/fisiología , Alimentación Animal , Animales , División Celular , Grasas de la Dieta/toxicidad , Femenino , Técnicas de Silenciamiento del Gen , Intolerancia a la Glucosa/fisiopatología , Hiperplasia , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Obesidad/etiología , Especificidad de Órganos , Ratas , Receptores de Somatotropina/antagonistas & inhibidores , Receptores de Somatotropina/genética , Factor de Transcripción STAT5/metabolismo
14.
Artículo en Inglés | MEDLINE | ID: mdl-22654802

RESUMEN

Growth hormone (GH), a master regulator of somatic growth, also regulates carbohydrate and lipid metabolism via complex interactions with insulin and insulin-like growth factor-1 (IGF-1). Data from human and rodent studies reveal the importance of GH in insulin synthesis and secretion, lipid metabolism and body fat remodeling. In this review, we will summarize the tissue-specific metabolic effects of GH, with emphasis on recent targets identified to mediate these effects. Furthermore, we will discuss what role GH plays in obesity and present possible mechanisms by which this may occur.

16.
Endocr Relat Cancer ; 17(4): 941-51, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20801951

RESUMEN

Type 2 diabetes increases breast cancer risk and mortality, and hyperinsulinemia is a major mediator of this effect. The mammalian target of rapamycin (mTOR) is activated by insulin and is a key regulator of mammary tumor progression. Pharmacological mTOR inhibition suppresses tumor growth in numerous mammary tumor models in the non-diabetic setting. However, the role of the mTOR pathway in type 2 diabetes-induced tumor growth remains elusive. Herein, we investigated whether the mTOR pathway is implicated in insulin-induced mammary tumor progression in a transgenic mouse model of type 2 diabetes (MKR mice) and evaluated the impact of mTOR inhibition on the diabetic state. Mammary tumor progression was studied in the double transgenic MMTV-Polyoma Virus middle T antigen (PyVmT)/MKR mice and by orthotopic inoculation of PyVmT- and Neu/ErbB2-driven mammary tumor cells (Met-1 and MCNeuA cells respectively). mTOR inhibition by rapamycin markedly suppressed tumor growth in both wild-type and MKR mice. In diabetic animals, however, the promoting action of insulin on tumor growth was completely blunted by rapamycin, despite a worsening of the carbohydrate and lipid metabolism. Taken together, pharmacological mTOR blockade is sufficient to abrogate mammary tumor progression in the setting of hyperinsulinemia, and thus mTOR inhibitors may be an attractive therapeutic modality for breast cancer patients with type 2 diabetes. Careful monitoring of the metabolic state, however, is important as dose adaptations of glucose- and/or lipid-lowering therapy might be necessary.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Insulina/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Apoptosis/fisiología , Glucemia/análisis , Peso Corporal/fisiología , Neoplasias de la Mama/patología , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Ingestión de Alimentos/fisiología , Femenino , Insulina/farmacología , Ratones , Ratones Transgénicos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Triglicéridos/sangre
17.
Mt Sinai J Med ; 77(2): 197-213, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20309918

RESUMEN

Epidemiological studies have demonstrated an association between type 2 diabetes and cancer. Type 2 diabetes is characterized by insulin resistance and hyperinsulinemia. Hyperinsulinemia may lead to cancer through insulin's effect on its cognate receptor and the insulin-like growth factor system. The effects of insulin and insulin-like growth factor I on cancer development and progression have been demonstrated in animal and human studies. Type 2 diabetes has been positively associated with cancers of the breast, colon, and pancreas. An inverse relationship has been observed between type 2 diabetes and prostate cancer, and this may be due to lower testosterone levels in men with type 2 diabetes. Medications used to treat type 2 diabetes may affect cancer cells directly or indirectly by affecting serum insulin levels. Hyperinsulinemia may be an important risk factor for cancer as well as a target for cancer therapy.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Neoplasias Colorrectales/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Neoplasias Pancreáticas/fisiopatología , Neoplasias de la Próstata/fisiopatología , Neoplasias de la Mama/epidemiología , Neoplasias Colorrectales/epidemiología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Progresión de la Enfermedad , Femenino , Humanos , Hiperinsulinismo , Insulina , Resistencia a la Insulina , Masculino , Neoplasias Pancreáticas/epidemiología , Neoplasias de la Próstata/epidemiología , Medición de Riesgo , Factores de Riesgo , Somatomedinas , Tiazolidinedionas/uso terapéutico
18.
Cancer Res ; 70(2): 741-51, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20068149

RESUMEN

Epidemiologic studies suggest that type 2 diabetes (T2D) increases breast cancer risk and mortality, but there is limited experimental evidence supporting this association. Moreover, there has not been any definition of a pathophysiological pathway that diabetes may use to promote tumorigenesis. In the present study, we used the MKR mouse model of T2D to investigate molecular mechanisms that link T2D to breast cancer development and progression. MKR mice harbor a transgene encoding a dominant-negative, kinase-dead human insulin-like growth factor-I receptor (IGF-IR) that is expressed exclusively in skeletal muscle, where it acts to inactivate endogenous insulin receptor (IR) and IGF-IR. Although lean female MKR mice are insulin resistant and glucose intolerant, displaying accelerated mammary gland development and enhanced phosphorylation of IR/IGF-IR and Akt in mammary tissue, in the context of three different mouse models of breast cancer, these metabolic abnormalities were found to accelerate the development of hyperplastic precancerous lesions. Normal or malignant mammary tissue isolated from these mice exhibited increased phosphorylation of IR/IGF-IR and Akt, whereas extracellular signal-regulated kinase 1/2 phosphorylation was largely unaffected. Tumor-promoting effects of T2D in the models were reversed by pharmacological blockade of IR/IGF-IR signaling by the small-molecule tyrosine kinase inhibitor BMS-536924. Our findings offer compelling experimental evidence that T2D accelerates mammary gland development and carcinogenesis,and that the IR and/or the IGF-IR are major mediators of these effects.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Hiperinsulinismo/metabolismo , Neoplasias Mamarias Experimentales/etiología , Neoplasias Mamarias Experimentales/metabolismo , Animales , Bencimidazoles/farmacología , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Femenino , Hiperinsulinismo/sangre , Hiperinsulinismo/patología , Insulina/sangre , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/sangre , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Transgénicos , Proteína Oncogénica v-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Piridonas/farmacología , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/antagonistas & inhibidores , Receptor de Insulina/metabolismo
19.
Diabetes ; 59(3): 686-93, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19959755

RESUMEN

OBJECTIVE: Type 2 diabetes increases breast cancer risk and mortality, and hyperinsulinemia has been identified as a major factor linking these two diseases. Thus, we hypothesized that pharmacological reduction of elevated insulin levels would attenuate type 2 diabetes-mediated mammary tumor progression. RESEARCH DESIGN AND METHODS: We studied mammary tumor development in MKR(+/+) mice, a nonobese, hyperinsulinemic mouse model of type 2 diabetes. MKR(+/+) mice were either crossed with mice expressing the polyoma virus middle T oncogene specifically in the mammary gland or inoculated orthotopically with the mouse mammary tumor cell lines Met-1 and MCNeuA. MKR(+/+) or control mice harboring tumors were treated with CL-316243, a specific beta3-adrenergic receptor agonist, which sensitizes insulin action but has no direct effect on the mouse mammary epithelium or Met-1 and MCNeuA cells. RESULTS: CL-316243 treatment significantly reduced the elevated insulin levels in MKR(+/+) mice and, as a consequence, attenuated mammary tumor progression in the three tumor models tested. This effect was accompanied by reductions in phosphorylation of insulin and IGF-I receptors in transformed mammary tissue. CONCLUSIONS: Insulin-sensitizing treatment is sufficient to abrogate type 2 diabetes-mediated mammary tumor progression. Therefore, early administration of insulin-sensitizing therapy may reduce breast cancer risk and mortality in patients with type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Dioxoles/farmacología , Hiperinsulinismo/tratamiento farmacológico , Hipoglucemiantes/farmacología , Neoplasias Mamarias Animales/tratamiento farmacológico , Animales , Composición Corporal/efectos de los fármacos , Línea Celular Tumoral , Comorbilidad , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/epidemiología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ingestión de Alimentos/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Femenino , Hiperinsulinismo/sangre , Hiperinsulinismo/epidemiología , Insulina/sangre , Lípidos/sangre , Masculino , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Animales/epidemiología , Neoplasias Mamarias Animales/patología , Ratones , Ratones Endogámicos , Ratones Transgénicos , Poliomavirus/genética , Factores de Riesgo
20.
Growth Horm IGF Res ; 20(1): 1-7, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19800274

RESUMEN

This review will summarize the metabolic effects of growth hormone (GH) on the adipose tissue, liver, and skeletal muscle with focus on lipid and carbohydrate metabolism. The metabolic effects of GH predominantly involve the stimulation of lipolysis in the adipose tissue resulting in an increased flux of free fatty acids (FFAs) into the circulation. In the muscle and liver, GH stimulates triglyceride (TG) uptake, by enhancing lipoprotein lipase (LPL) expression, and its subsequent storage. The effects of GH on carbohydrate metabolism are more complicated and may be mediated indirectly via the antagonism of insulin action. Furthermore, GH has a net anabolic effect on protein metabolism although the molecular mechanisms of its actions are not completely understood. The major questions that still remain to be answered are (i) What are the molecular mechanisms by which GH regulates substrate metabolism? (ii) Does GH affect substrate metabolism directly or indirectly via IGF-1 or antagonism of insulin action?


Asunto(s)
Metabolismo de los Hidratos de Carbono/fisiología , Hormona del Crecimiento/fisiología , Metabolismo de los Lípidos/fisiología , Tejido Adiposo/metabolismo , Animales , Femenino , Humanos , Resistencia a la Insulina/fisiología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Hígado/metabolismo , Masculino , Ratones , Músculo Esquelético/metabolismo , Proteínas/metabolismo , Ratas , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...